Β-Catenin Stabilization in Skin Fibroblasts Causes Fibrotic Lesions by Preventing Adipocyte Differentiation of the Reticular Dermis  Maria Mastrogiannaki,

Slides:



Advertisements
Similar presentations
Volume 9, Issue 5, Pages (November 2017)
Advertisements

Involvement of Wnt Signaling in Dermal Fibroblasts
Blockade of PDGF Receptors by Crenolanib Has Therapeutic Effect in Patient Fibroblasts and in Preclinical Models of Systemic Sclerosis  Katsunari Makino,
Targeted Disruption of the Lama3 Gene in Adult Mice Is Sufficient to Induce Skin Inflammation and Fibrosis  Monika Pesch, Sabrina König, Monique Aumailley 
PFKFB3, a Direct Target of p63, Is Required for Proliferation and Inhibits Differentiation in Epidermal Keratinocytes  Robert B. Hamanaka, Gökhan M. Mutlu 
Ceramide Synthase 4 Regulates Stem Cell Homeostasis and Hair Follicle Cycling  Franziska Peters, Susanne Vorhagen, Susanne Brodesser, Kristin Jakobshagen,
Glycoprotein Nonmelanoma Clone B Regulates the Crosstalk between Macrophages and Mesenchymal Stem Cells toward Wound Repair  Bing Yu, Talib Alboslemy,
Emily J. Hamburg, Radhika P. Atit  Journal of Investigative Dermatology 
S100A12 Induced in the Epidermis by Reduced Hydration Activates Dermal Fibroblasts and Causes Dermal Fibrosis  Jingling Zhao, Aimei Zhong, Emily E. Friedrich,
Sarah A. Best, Amy N. Nwaobasi, Chrysalyne D. Schmults, Matthew R
Β-Catenin Stabilization in Skin Fibroblasts Causes Fibrotic Lesions by Preventing Adipocyte Differentiation of the Reticular Dermis  Maria Mastrogiannaki,
C-Myc activation in transgenic mouse epidermis results in mobilization of stem cells and differentiation of their progeny  Isabel Arnold, Fiona M Watt 
Volume 13, Issue 4, Pages (October 2013)
Sarah A. Best, Amy N. Nwaobasi, Chrysalyne D. Schmults, Matthew R
Tbx18 Targets Dermal Condensates for Labeling, Isolation, and Gene Ablation during Embryonic Hair Follicle Formation  Laura Grisanti, Carlos Clavel, Xiaoqiang.
Kai Kretzschmar, Denny L. Cottle, Pawel J. Schweiger, Fiona M. Watt 
Jiang Chen, Christine Laclef, Alejandra Moncayo, Elizabeth R
HPV8 Field Cancerization in a Transgenic Mouse Model Is due to Lrig1+ Keratinocyte Stem Cell Expansion  Simone Lanfredini, Carlotta Olivero, Cinzia Borgogna,
Fibroblast-Derived MMP-14 Regulates Collagen Homeostasis in Adult Skin
James M. Waters, Jessica E. Lindo, Ruth M. Arkell, Allison J. Cowin 
Isolation of Murine Hair-Inducing Cells Using the Cell Surface Marker Prominin- 1/CD133  Yuriko Ito, Tatsuo S. Hamazaki, Kiyoshi Ohnuma, Kunihiko Tamaki,
Makoto Takeo, Christopher S. Hale, Mayumi Ito 
Keratinocyte-Derived Chemokines Orchestrate T-Cell Positioning in the Epidermis during Vitiligo and May Serve as Biomarkers of Disease  Jillian M. Richmond,
Patched Receptors Sense, Interpret, and Establish an Epidermal Hedgehog Signaling Gradient  Christelle Adolphe, Jan Philipp Junker, Anna Lyubimova, Alexander.
Atypical Protein Kinase C Isoform, aPKCλ, Is Essential for Maintaining Hair Follicle Stem Cell Quiescence  Shin-Ichi Osada, Naoko Minematsu, Fumino Oda,
Activating Hair Follicle Stem Cells via R-spondin2 to Stimulate Hair Growth  Andrew A. Smith, Jingtao Li, Bo Liu, Daniel Hunter, Malcolm Pyles, Martin.
Enrichment for Living Murine Keratinocytes from the Hair Follicle Bulge with the Cell Surface Marker CD34  Rebecca J. Morris, Carl D. Bortner, George.
Transcription Factor MafB Coordinates Epidermal Keratinocyte Differentiation  Masashi Miyai, Michito Hamada, Takashi Moriguchi, Junichiro Hiruma, Akiyo.
Identification of a Potential Molecular Diagnostic Biomarker in Keloid Disease: Syndecan-1 (CD138) Is Overexpressed in Keloid Scar Tissue  Rania A. Bagabir,
Christina A. Young, Richard L
Thy-1/β3 Integrin Interaction-Induced Apoptosis of Dermal Fibroblasts Is Mediated by Up-Regulation of FasL Expression  Manuela Schmidt, Danny Gutknecht,
Nicole Amberg, Martin Holcmann, Gabriel Stulnig, Maria Sibilia 
Peggy S. Myung, Makoto Takeo, Mayumi Ito, Radhika P. Atit 
Volume 9, Issue 5, Pages (November 2017)
Role of the Notch Ligand Delta1 in Embryonic and Adult Mouse Epidermis
Integrin β6-Deficient Mice Show Enhanced Keratinocyte Proliferation and Retarded Hair Follicle Regression after Depilation  Yanshuang Xie, Kevin J. McElwee,
Transcription Factor CTIP2 Maintains Hair Follicle Stem Cell Pool and Contributes to Altered Expression of LHX2 and NFATC1  Shreya Bhattacharya, Heather.
Fuz Controls the Morphogenesis and Differentiation of Hair Follicles through the Formation of Primary Cilia  Daisy Dai, Huiping Zhu, Bogdan Wlodarczyk,
Volume 16, Issue 2, Pages (July 2016)
Sustained Activation of Fibroblast Transforming Growth Factor-β/Smad Signaling in a Murine Model of Scleroderma  Shinsuke Takagawa, Gabriella Lakos, Yasuji.
Gorab Is Required for Dermal Condensate Cells to Respond to Hedgehog Signals during Hair Follicle Morphogenesis  Ying Liu, Elizabeth R. Snedecor, Yeon.
Alexandra Charruyer, Lauren R. Strachan, Lili Yue, Alexandra S
Overexpression of CD109 in the Epidermis Differentially Regulates ALK1 Versus ALK5 Signaling and Modulates Extracellular Matrix Synthesis in the Skin 
Slc1a3-CreER as a Targeting Tool for the K6+ Epithelial Stem Cell Niche and its Precursors during Mouse Hair Follicle Cycle  Aiko Sada, Prachi Jain, Sherry.
Collagen VII Half-Life at the Dermal-Epidermal Junction Zone: Implications for Mechanisms and Therapy of Genodermatoses  Tobias Kühl, Markus Mezger, Ingrid.
Lineage Identity and Location within the Dermis Determine the Function of Papillary and Reticular Fibroblasts in Human Skin  Ana Korosec, Sophie Frech,
14-3-3σ Regulates Keratinocyte Proliferation and Differentiation by Modulating Yap1 Cellular Localization  Sumitha A.T. Sambandam, Ramesh B. Kasetti,
Different Consequences of β1 Integrin Deletion in Neonatal and Adult Mouse Epidermis Reveal a Context-Dependent Role of Integrins in Regulating Proliferation,
Keratinocyte-Specific Deletion of the Receptor RAGE Modulates the Kinetics of Skin Inflammation In Vivo  Julia S. Leibold, Astrid Riehl, Jan Hettinger,
Modulation of Hair Growth with Small Molecule Agonists of the Hedgehog Signaling Pathway  Rudolph D. Paladini, Jacqueline Saleh, Changgeng Qian, Guang-Xin.
CD301b+ Macrophages Are Essential for Effective Skin Wound Healing
The Vitamin D Receptor Is Required for Mouse Hair Cycle Progression but not for Maintenance of the Epidermal Stem Cell Compartment  Héctor G. Pálmer,
Dual Role of the Anaphase Promoting Complex/Cyclosome in Regulating Stemness and Differentiation in Human Primary Keratinocytes  Ling Shih Quek, Nicolas.
Yuko Oda, Lizhi Hu, Vadim Bul, Hashem Elalieh, Janardan K
Volume 25, Issue 11, Pages (November 2017)
Transient Expression of Ephrin B2 in Perinatal Skin Is Required for Maintenance of Keratinocyte Homeostasis  Gyohei Egawa, Masatake Osawa, Akiyoshi Uemura,
Fate of Prominin-1 Expressing Dermal Papilla Cells during Homeostasis, Wound Healing and Wnt Activation  Grace S. Kaushal, Emanuel Rognoni, Beate M. Lichtenberger,
Epidermal Stem Cells in the Isthmus/Infundibulum Influence Hair Shaft Differentiation: Evidence from Targeted DLX3 Deletion  Jin-Chul Kim, Olivier Duverger,
Andreya Sharov, Desmond J. Tobin, Tatyana Y
The Nf1 Tumor Suppressor Regulates Mouse Skin Wound Healing, Fibroblast Proliferation, and Collagen Deposited by Fibroblasts  Radhika P. Atit, Maria J.
Increased Expression of Wnt2 and SFRP4 in Tsk Mouse Skin: Role of Wnt Signaling in Altered Dermal Fibrillin Deposition and Systemic Sclerosis  Julie Bayle,
Jaana Mannik, Kamil Alzayady, Soosan Ghazizadeh 
The EGFR Is Required for Proper Innervation to the Skin
Epidermal Nerve Fibers Modulate Keratinocyte Growth via Neuropeptide Signaling in an Innervated Skin Model  Dennis Roggenkamp, Sarah Köpnick, Franz Stäb,
Colin A.B. Jahoda, Adam C. Gilmore 
Expression of Activated MEK1 in Differentiating Epidermal Cells Is Sufficient to Generate Hyperproliferative and Inflammatory Skin Lesions  Robin M. Hobbs,
Yap Controls Stem/Progenitor Cell Proliferation in the Mouse Postnatal Epidermis  Annemiek Beverdam, Christina Claxton, Xiaomeng Zhang, Gregory James,
IL-17A Upregulates Keratin 17 Expression in Keratinocytes through STAT1- and STAT3- Dependent Mechanisms  Xiaowei Shi, Liang Jin, Erle Dang, Ting Chang,
Remodelling of adult dermis originates from a population of fibroblasts in the hair follicle junctional zone. Remodelling of adult dermis originates from.
DLX3-Dependent STAT3 Signaling in Keratinocytes Regulates Skin Immune Homeostasis  Shreya Bhattacharya, Jin-Chul Kim, Youichi Ogawa, Gaku Nakato, Veronica.
Presentation transcript:

β-Catenin Stabilization in Skin Fibroblasts Causes Fibrotic Lesions by Preventing Adipocyte Differentiation of the Reticular Dermis  Maria Mastrogiannaki, Beate M. Lichtenberger, Andreas Reimer, Charlotte A. Collins, Ryan R. Driskell, Fiona M. Watt  Journal of Investigative Dermatology  Volume 136, Issue 6, Pages 1130-1142 (June 2016) DOI: 10.1016/j.jid.2016.01.036 Copyright © 2016 The Authors Terms and Conditions

Figure 1 Localization and isolation of Sca1+ and Sca1- dermal fibroblasts. (a–d, n–q) Sections of P2 back skin from PdgfrαEGFP mice showing expression of nuclear EGFP and immunostaining for (a, b) K14 and PDGFRα, (c, d) Sca1, (n, o) Fabp4, and (p, q) Perilipin. (b, d, o, q) are enlargements of selected regions of (a, c, n, p), respectively. Scale bars = 150 μm. (e) Flow cytometry plot showing gating of different dermal subpopulations on the basis of PdgfrαEGFP and Sca1 expression. (f–m) Quantitative real-time PCR analysis of mRNA levels in sorted cell populations: PdgfrαEGFP-/Sca1-, PdgfrαEGFP+/Sca1- and PdgfrαEGFP+/Sca1+. Genes were normalized to the Gapdh gene. Error bars represent standard error of the mean of replicates from four mice. ∗P ≤ 0.05, ∗∗P ≤ 0.005, ∗∗∗P ≤ 0.0005 compared with GFP- cells; #p ≤ 0.05 compared with EGFP+/Sca1- cells. ad, adipocyte; epi, epidermis; der, dermis; EGFP, enhanced green fluorescent protein; GFP, green fluorescent protein; HF, hair follicle; pap, papillary; PDGFRα, platelet-derived growth factor receptor α; ret, reticular. Journal of Investigative Dermatology 2016 136, 1130-1142DOI: (10.1016/j.jid.2016.01.036) Copyright © 2016 The Authors Terms and Conditions

Figure 2 Distinct transcriptional signature of PdgfrαEGFP+/Sca1+ dermal cells. (a) Heat map showing hierarchical clustering (based on entities and samples) of all differentially regulated genes (P < 0.05, change >2-fold) between PdgfrαEGFP+/Sca1+ and PdgfrαEGFP+/Sca1- fibroblasts. (b) Selected genes up-regulated or down-regulated in Sca1+ cells. Values in parentheses represent fold change of each gene. (c) Heat map showing hierarchical clustering (based on entities) of all regulated genes in the Gene Ontology term “Wnt receptor signaling pathway”. (d–k) Quantitative real-time PCR analysis of mRNA levels in sorted cell populations, normalized to Gapdh gene expression. Error bars represent standard error of the mean of replicates from four mice. ∗P ≤ 0.05, ∗∗P ≤ 0.005, ∗∗∗P ≤ 0.0005 compared with GFP- cells; #P ≤ 0.05 compared with GFP+/Sca1- cells. (l, m) Immunofluorescent staining of neonatal skin with an antibody detecting β-catenin. Red arrowheads show β-catenin+ fibroblasts in the reticular dermis. 4′, 6-diamidino-2-phenylindole labels nuclei. Scale bar = 200 μm. (n) Section of P1 back skin immunostained for Tcf3/4 (red) and Lef1 (green). White arrowheads indicate double-labeled cells. Dashed lines demarcate epidermal-dermal boundary. Scale bar = 100 μm. (o–r) Higher-magnification images of the boxed areas in (m, n), showing upper (o, q) and lower (p, r) dermis. BMP, bone morphogenic protein; DAPI, 4′, 6-diamidino-2-phenylindole; GO, Gene Ontology; PDGF, platelet-derived growth factor. Journal of Investigative Dermatology 2016 136, 1130-1142DOI: (10.1016/j.jid.2016.01.036) Copyright © 2016 The Authors Terms and Conditions

Figure 3 β-Catenin stabilization in dermal fibroblasts via PdgfrαCreERT2. (a) Schematic illustration of experimental strategy. (b) Recombination efficiency of PDGFRαCreERT2. Bar graph showing percentage of tdTomato+ cells within different fibroblast subsets after gating for ITGA6-/LIN-/CD31- cells 2 days after 4-OHT–mediated recombination. Data are reported as mean ± standard error of the mean of triplicate samples in a representative experiment (n = 2–3 independent experiments). (c, d) tdTomato expression in 4-OHT–treated dorsal skin from (c) PDGFRαCreERT2 × tdTomato-LSL × Ctnnb1 Exon3+/+ (control) and (d) PDGFRαCreERT2 × tdTomato-LSL × Ctnnb1 Exon3Flox/+ (cΔex3) mice at P4. (e–h) Strong β-catenin staining is detected in tdTomato+ fibroblasts of mice with activated Wnt/β-catenin signaling. Note that the PDGFRαCreERT2 transgene is also active in cells of the dermal papilla (white arrowheads in g). Red arrow heads in (h) depict β-catenin+ cells in the dermal papilla. (i–p) Paraffin sections of back skin of control and mutant (cΔex3) littermates stained for hematoxylin and eosin at (i, j) P4, (k, l) first telogen (P18), (m, n) anagen (P35), and (o, p) second telogen (P56). Boxed areas in (k, l) are shown as higher-magnification inserts. Scale bars = 200 μm. (q, r) Hair follicle length measured in (q) male and (r) female cΔex3 and control mice at P4, P11, P18, P24, P28, P35, P42, and P56 (n = 2–8). Data points are reported as mean ± standard error of the mean. (s) Quantification of proliferating, phosphohistone H3+ cells in the outer root sheath of hair follicles in immunostained skin sections (n = 3 biological samples; ≥5 hair follicles per biological sample were scored). 4-OHT, 4-hydroxy-tamoxifen; ctr, control; Dapi, 4′, 6-diamidino-2-phenylindole; K14, keratin 14. Journal of Investigative Dermatology 2016 136, 1130-1142DOI: (10.1016/j.jid.2016.01.036) Copyright © 2016 The Authors Terms and Conditions

Figure 4 Induction of dermal fibrosis and proliferation by β-catenin stabilization. (a, b) Herovici’s staining differentiates between immature collagen fibers (blue) versus mature collagen (pink). Note fibrotic region with mature extracellular matrix in (b) (boxed). (c) Relative expression of adipocyte differentiation genes in flow-sorted tdtomato+ fibroblasts. Data represent mean ± standard error of the mean (n = 3). ∗P ≤ 0.05. (d–i) Immunostained paraffin sections of 35-day-old mutant and control skin. (j, k) Whole-mount thick sections stained with LipidTox (green) with DAPI counterstain (blue). (l, m) Quantification in paraffin sections of P35 skin of (l) adipocytes and (m) EdU+ fibroblasts in the adipocyte layer and interfollicular dermis (including papillary fibroblasts, dermal sheath, and reticular fibroblasts). The boundary between papillary/reticular and adipocyte layers was defined by the interface between the bottom of the reticular layer and the upper layer of differentiated adipocytes. Data points represent mean ± standard error of the mean; n ≥ 5 for each group. ∗P ≤ 0.05; ∗∗P ≤ 0.005; ∗∗∗P ≤ 0.0005. (n, o) Skin labeled with EdU (green) for 4 hours before harvesting, costained for Pdgfrα (red), keratin 14 (white), and DAPI (blue). White arrowheads indicate EdU+ fibroblasts within the adipose layer. Inserts are higher-magnification images of the boxed areas. Scale bars = 200 μm. ctr, control; DAPI, 4′, 6-diamidino-2-phenylindole; K14, keratin 14. Journal of Investigative Dermatology 2016 136, 1130-1142DOI: (10.1016/j.jid.2016.01.036) Copyright © 2016 The Authors Terms and Conditions

Figure 5 β-Catenin stabilization in the lower dermis via Dlk1CreERT2. (a) Schematic illustration of experimental strategy. (b, c) Quantification of tdTomato+ cells in E18.5 dermal cell suspensions (ITGA6-/LIN-/CD31-) 2 days after 4-OHT injection. (b) Percentage of tdTomato+ cells in each fibroblast subpopulation. (c) Contribution of each fibroblast subpopulation to tdTomato+ fibroblasts. Data are presented as mean ± standard error of the mean of quadruplicate samples in a representative experiment (n = 3 independent experiments). (d, e) tdTomato-expressing cells in P56 back skin of Dlk1CreERT2 × tdTomato-LSL × Ctnnb1 Exon3+/+ (control) and Dlk1CreERT2 × tdTomato-LSL × Ctnnb1 Exon3Flox/+ (Dlk1LTΔex3) mice. Asterisks in (d) indicate terminally differentiated adipocytes. (f, g) Hematoxylin and eosin and (h, i) Herovici’s staining of paraffin sections of P56 control and Dlk1LTΔex3 mouse back skin. Arrows in (g) and (i) indicate fibrotic regions. (j–m) P56 control and Dlk1LTΔex3 back skin labeled for β-catenin or caveolin-1 and keratin 14, counterstained with DAPI. Vertical lines indicate thickness of adipocyte layer. Dashed line in (k) demarcates fibrotic tissue (FT). (n, o) Quantification of fibrotic area shown as percentage of the adipocyte layer (n) and number of adipocytes (o). (p) Quantification of EdU-labeled fibroblasts in the interfollicular dermis (including papillary fibroblasts, dermal sheath, and reticular fibroblasts) and adipose layer (defined as in Figure 4m) after a 2-hour EdU pulse. Data are presented as mean ± standard error of the mean of triplicate samples in a representative experiment (n = 3–5). ∗P ≤ 0.05; ∗∗P ≤ 0.005. (q, r) EdU staining of sections shown in (d) and (e). Scale bars = 200 μm. 4-OHT, 4-hydroxy-tamoxifen; AL, adipocyte layer; ctr, control; DAPI, 4′, 6-diamidino-2-phenylindole; K14, keratin 14. Journal of Investigative Dermatology 2016 136, 1130-1142DOI: (10.1016/j.jid.2016.01.036) Copyright © 2016 The Authors Terms and Conditions

Figure 6 Effect of β-catenin stabilization on adipocyte differentiation in cultured Sca1+ fibroblasts. (a) Schematic illustration of experimental strategy. (b–d) Sca1+/tdTomato+ targeted fibroblasts from dermal cell suspensions of PDGFRαCreERT2 × tdTomato-LSL × Ctnnb1 Exon3+/+ (control) and PDGFRαCreERT2 × tdTomato-LSL × Ctnnb1 Exon3Flox/+ (cΔex3) littermates were isolated by flow cytometry. (b, c) Gating out α6 integrin-positive cells (ITGA6; keratinocytes) (b) and positive selection for Sca1+/tdTomato+ cells (c). (d) Recombination efficiency (percentage tdTomato+ cells) in Cd24+/Sca1- and Cd24-/Sca1+ cells. Data are presented as mean ± standard error of the mean of triplicate samples in a representative experiment (n = 3 independent experiments). (e) Examples of individual colonies formed by Sca1+ fibroblasts stained for LipidTox (green), tdTomato (red), and DAPI (blue). Scale bars = 10 μm. (f, g) Percentage of colonies containing 1, 2, 3–5, or more than 5 cells in standard (f) and adipogenic (g) medium. (h, i) Total LipidTox fluorescence (lipid intensity) per clone in control and mutant (cΔex3) cultures grown in standard (h) or adipogenic (i) medium. (f–i) n = 2–3 biological replicates and 2 technical replicates. ∗P ≤ 0.05, ∗∗P ≤ 0.01, ∗∗∗P ≤ 0.005. (f, g) two-way analysis of variance with a Bonferroni posttest. (h, i) 25% confidence intervals are shown. 4-OHT, 4-hydroxy-tamoxifen; ctr, control DAPI, 4′, 6-diamidino-2-phenylindole; FSC, forward scatter; ns, difference not significant. Journal of Investigative Dermatology 2016 136, 1130-1142DOI: (10.1016/j.jid.2016.01.036) Copyright © 2016 The Authors Terms and Conditions